investigating the molecular mechanisms of neisseria...

129
Alma Mater Studiorum – Università di Bologna DOTTORATO DI RICERCA IN BIOLOGIA CELLULARE E MOLECOLARE Ciclo XXIX Settore Concorsuale SSC 05/E2 Settore Disciplinare SSD BIO/11 Investigating the molecular mechanisms of Neisseria meningitidis antigen regulation: determining a switch between colonization and invasion Presentata da: Sara Borghi Coordinatore Dottorato Prof. Giovanni Capranico Relatore: Prof. Vincenzo Scarlato Correlatore: Dott.ssa Isabel Delany Esame finale anno 2017

Upload: others

Post on 22-May-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

  • Alma Mater Studiorum – Università di Bologna

    DOTTORATO DI RICERCA IN

    BIOLOGIA CELLULARE E MOLECOLARE

    Ciclo XXIX

    Settore Concorsuale SSC 05/E2

    Settore Disciplinare SSD BIO/11

    Investigating the molecular mechanisms of Neisseria meningitidis antigen regulation:

    determining a switch between colonization and invasion

    Presentata da: Sara Borghi

    Coordinatore Dottorato

    Prof. Giovanni Capranico

    Relatore:

    Prof. Vincenzo Scarlato

    Correlatore:

    Dott.ssa Isabel Delany

    Esame finale anno 2017

  • 2

    Table of Contents

    1. Abstract ........................................................................................................................................ 4

    2. Introduction ................................................................................................................................. 6

    2.1 Meningococcal disease ......................................................................................................... 6

    2.2 Neisseria meningitidis: pathogen and pathogenesis ........................................................... 7

    2.3 Meningococcal virulence factors ....................................................................................... 11

    2.4 Meningococcal vaccines ..................................................................................................... 17

    2.4.1 Licensed vaccines against MenB ................................................................................. 18

    2.4.2 Investigational MenB vaccines .................................................................................... 20

    Chapter 1: NHBA regulation and expression during colonization and invasion

    Sensing the environment .......................................................................................................................... 24

    Neisserial Heparin Binding Antigen (NHBA) ...................................................................................... 25

    3. Results ........................................................................................................................................ 28

    3.1 NHBA expression and surface exposure are temperature-dependent ........................ 28

    3.2 Mutations and deletions in the 5’UTR and 5’TR of nhba affect expression ................ 31

    3.3 NHBA is expressed during the active growth ................................................................ 38

    3.4 NHBA thermoregulation is not driven by the nhba promoter ...................................... 38

    3.5 Temperature affetcs nhba RNA half-life ........................................................................... 41

    3.6 NHBA protein shows higher stability at 30°C respect to 37°C ..................................... 44

    3.7 NHBA expression levels correlate with susceptibility to complement-mediated killing by anti-NHBA antibodies ............................................................................................... 45

    3.8 NHBA regulation during invasion ................................................................................... 50

    3.9 NHBA cleavage does not affect NHBA-mediated killing suceptibility ...................... 54

    4. Discussion and conclusions ................................................................................................... 56

    Chapter 2: Comparing different delivery systems by using NadA as a model antigen

    Immune response and vaccine design ................................................................................................... 67

    Neisserial adhesin A (NadA) ................................................................................................................... 69

    5. Results ........................................................................................................................................ 72

    5.1 NadA overexpression on MenB and E.coli nOMV ......................................................... 72

    5.2 Prototype nOMV vaccines elicited higher titre of α-NadA antibodies ....................... 74

    5.3 Evaluation of the bactericidal activity of the antibodies elicited through rSBA assay ............................................................................................................................................... 76

    5.4 Inhibition of E.coli-NadA var3 adhesion to Chang epithelial cells with different sera .................................................................................................................................................. 79

  • 3

    6. Discussion and conclusions ................................................................................................... 81

    7. Materials and methods ............................................................................................................ 86

    7.1 Bacterial strains and culture conditions ........................................................................... 86

    7.2 Generation of plasmids and N.meningitidis new recombinant strains ....................... 86

    7.3 Polyacrylamide gel electrophoresis and Western blotting ............................................ 88

    7.4 Quantitative real-time PCR (qRT-PCR) experiments ..................................................... 88

    7.5 Flow cytometry .................................................................................................................... 89

    7.6 Serum Bactericidal Assay (SBA) ........................................................................................ 90

    7.7 RNA stability assay ............................................................................................................. 90

    7.8 Protein stability assay ......................................................................................................... 90

    7.9 nOMV preparation .............................................................................................................. 91

    7.10 Transmission Electron Microscopy ................................................................................. 92

    7.11 Mice immunizations ......................................................................................................... 93

    7.12 IgG antibody titers elicited against NadA ..................................................................... 93

    7.13 Inhibition of the binding assay ........................................................................................ 94

    8. Appendix .................................................................................................................................... 95

    9. References ................................................................................................................................ 104

  • 4

    1. Abstract

    Neisseria meningitidis colonizes the nasopharynx of humans and pathogenic strains can

    disseminate into the bloodstream causing septicemia and meningitis. Neisserial

    Heparin Binding Antigen (NHBA) and Neisserial Adhesin A (NadA) are part of a

    multicomponent vaccine against N. meningitidis serogroup B, called 4CMenB

    (Bexsero™).

    NHBA is a surface-exposed lipoprotein which is expressed by all N. meningitidis strains

    in different isoforms. NHBA harbors an arginine-rich motif through which it is able to

    bind heparin-like molecules, increasing adherence to host tissues and heparin-

    mediated serum resistance. We determined that temperature controlled the expression

    of NHBA in all strains tested, regardless of the clonal complex or peptide isoform

    expressed. NHBA expression was significantly increased at 30-32°C compared to 37°C,

    the temperature standardly used for in vitro culturing. An increase in NHBA

    expression at lower temperatures was measurable both at protein and RNA levels and

    was also reflected by a higher surface exposure of this antigen. A detailed molecular

    analysis indicated that multiple molecular mechanisms are responsible for the

    thermoregulated NHBA expression. The comparison of RNA steady state levels in cells

    cultured at 30°C and 37°C demonstrated an increased RNA stability/translatability at

    lower temperatures. Furthermore, protein stability was also impacted resulting in

    higher NHBA stability at lower temperatures. Increased NHBA expression resulted in

    more efficient killing as shown by serum bactericidal assay (SBA). Mimicking the

    invasive condition, we investigated the NHBA expression in response to the presence

    of serum. We showed that the presence of human serum has contrasting effects on

    NHBA expression, resulting in transient up-regulation of NHBA at transcriptional

    level, however the protein is rapidly processed likely by complement proteases. We

    propose a model in which NHBA regulation in response to temperature downshift

    might reflect the bacterial adaptation during the initial step of host-bacterial interaction

    and might also explain higher susceptibility to anti-NHBA antibodies in the

    nasopharynx niche. On the other hand, the initial up-regulation and the high

    processing of NHBA might play a role during the first steps of invasive disease. All

    together these data describe the importance of NHBA both as virulence factor and as

    vaccine antigen during neisserial colonization and invasion.

  • 5

    In the second part of the thesis, we compared genetically engineered outer membrane

    vesicles and recombinant proteins, as delivery systems of protective antigen. Using

    NadA as model antigen, we determined that OMV overexpressing NadA produced by

    homologous (MenB) or heterologous (E.coli) bacterial strains, are able to elicit a higher

    functional antibody response respect to the recombinant protein per se, despite the

    comparable anti-NadA titres elicited. The differences in functionality might be due to

    different IgG subclasses distribution. Moreover, OMV overexpressing NadA are able to

    elicit antibodies that inhibit NadA-mediated adhesion on the host cells surface, in a

    much more efficient way respect to the recombinant protein formulation.

    These results indicate that the antigen delivered on OMV triggers a good functional

    immune response. This preliminary characterization supports the use of OMV as

    delivery systems for next generation vaccine design and remark the great potential of

    NadA as model antigen.

  • 6

    2. Introduction

    2.1. Meningococcal disease

    Invasive meningococcal disease is characterized by a rapid onset and progression to

    meningitis and/or sepsis which can lead to death within hours. The etiological agent of

    this devastating disease is Neisseria meningitidis, otherwise known as meningococcus.

    The first report of meningococcal disease is dated back to 1887 by Anton

    Weichselbaum, who described the meningococcal infection of the cerebrospinal fluid of

    a patient (Weichselbaum, A. 1887). Each year there are an estimated 1.2 million cases of

    invasive meningococcal disease and 135,000 deaths (WHO 2010). Despite the

    availability of antibiotic treatment, approximately 10 to 14% of people who contract

    meningococcal disease die, and the rate increases to 40-55% in the case of sepsis

    (Brandtzaeg, P. et al. 2005, Rosenstein, N.E. et al. 2001). Furthermore, approximately 11

    to 19% of individuals surviving the disease often suffer from permanent sequelae,

    including neuro-developmental deficits, hearing loss, ataxia, hemiplegia, seizures and

    limbs loss (Kaplan, S.L. et al. 2006, Rosenstein, N.E. et al. 2001, Thompson, M.J. et al.

    2006, WHO 2010).

    Multiple studies have demonstrated that carriage rates are very low in the first few

    years of life, but rise during adolescence, reaching peaks of 10-35%, and decreasing to

    less than 10% in older groups (Caugant, D.A. et al. 2007, Claus, H. et al. 2005). In

    contrast to carriage rates, meningococcal disease is rare, varying from 0.5 to 10 per

    100,000 persons; however, the incidence can rise above 1 per 1,000 persons during

    epidemics (Caugant, D.A. et al. 2009, Stephens, D.S. et al. 2007). Most cases of

    meningococcal disease occur in otherwise healthy individuals without identified risk

    factors and what determines the transition from colonization to invasive disease is not

    yet fully understood. However, certain biological, environmental and social factors

    have been associated with an increased risk of disease. Infants under one year of age

    have the highest risk of infection due to their immature immune systems (6.33-7.08

    cases per 100,000). Whereas, the peak observed in adolescents is largely due to

    increased carriage in this population (Cohn, A.C. et al. 2010). Several studies

    demonstrated that both host and pathogen factors influence the development of the

    disease, such as human genetic polymorphisms, impaired immune system, microbial

    virulence factors, as well as environmental conditions facilitating exposure and

  • 7

    acquisition, and naso- and oro-pharyngeal irritation caused by smoking and

    respiratory tract infection (Brigham, K.S. et al. 2009, Davila, S. et al. 2010,

    Goldschneider, I. et al. 1969, Harrison, L.H. 2006, Imrey, P.B. et al. 1995, Rosenstein,

    N.E. et al. 2001, Zuschneid, I. et al. 2008). The unspecific symptoms at the onset and the

    early stage of infection, like headache, fever and rash, can implicate an arduous

    diagnosis. Due to the rapid progression of this life-threatening pathology, vaccination

    represents the unique effective public health response.

    2.2. Neisseria meningitidis: pathogen and pathogenesis

    N. meningitidis is a strictly human, Gram-negative β-proteobacterium member of the

    Neisseriaceae family. It is an aerobic, non-motile and non-sporulating diplococcus

    (Figure 2.1), usually encapsulated and piliated.

    Figure 2.1. Immuno-gold labelling and transmission electron microscopy of Neisseria

    meningitidis. Analysis of the strain was performed with antisera raised against the NadA

    adhesin. Scale bars: 200 nm (from (Pizza, M. et al. 2000)

    The envelope of N. meningitidis consists of the cytoplasmic membrane, the outer

    membrane (OM) and the periplasm between them, which contains a layer of

    peptidoglycan. The cytoplasmic membrane is a phospholipid bilayer, whereas the OM

    is composed of a phospholipidic inner leaflet and an outer leaflet of

    lipooligosaccharide (LOS). Some meningococcal strains have a polysaccharide capsule

  • 8

    attached to their OM and almost all pathogenic strains are encapsulated. Nevertheless,

    also non-encapsulated isolates have been recently associated to invasive disease

    (Johswich, K.O. et al. 2012). On the basis of the bacterial polysaccharide capsule, N.

    meningitidis can be classified into at least thirteen serogroups: A, B, C, E-29, H, I, K, L,

    W, X, Y, Z and 29E (Branham, S.E. 1953). Among them, six serogroups (A, B, C, X, Y

    and W) are responsible for more than 90% of meningococcal disease worldwide and

    are thus considered pathogenic (Boisier, P. et al. 2007, Frasch, C.E. 1989, Jarvis, G.A. et

    al. 1987, Stephens, D.S. et al. 2007). Meningococci are further classified into serotypes

    and serosubtypes according to antigenic differences in their major outer membrane

    proteins, PorA and PorB. However, the classification based on the serological

    characteristics of N. meningitidis is limited due to the high frequency of variation of

    OM-proteins, probably determined by a strong selective pressure. Hence, new DNA-

    based methods for the characterization of meningococcal isolates have been developed,

    and the Multi Locus Sequence Typing (MLST) is now considered the gold standard for

    molecular typing and epidemiologic studies (Maiden, M.C. et al. 1998). This typing

    system relies on polymorphisms within seven housekeeping genes; each sequence for a

    given locus is screened for identity with already known sequences for that locus. If the

    sequence is different, it is considered to be a new allele and an identification number is

    assigned. Therefore, the combination of the seven allele numbers determines the allelic

    profile of the strain, and each different allelic profile is assigned as a sequence type

    (ST). Meningococci sharing at least four of the seven loci with a central ancestral

    genotype are grouped together into clonal complexes (CCs) (Urwin, R. et al. 2003).

    Through the employment of MLST it has been shown that the majority of strains

    associated with invasive disease belong to specific CCs (ST-1, ST-4, ST-5, ST-8, ST-11,

    ST-32, ST41/44 and ST-269), called hyper-invasive (Caugant, D.A. 2008, Maiden, M.C.

    2008). However, the reasons of this enhanced pathogenic phenotype are yet unknown.

  • 9

    The pathogenesis of N. meningitidis is a complex multi-stage process (Figure 2.2).

    Figure 2.2 Stages in the pathogenesis of Neisseria meningitidis. N. meningitidis may be

    acquired through the inhalation of respiratory droplets. The organism establishes intimate

    contact with non-ciliated mucosal epithelial cells of the upper respiratory tract, where it may

    enter the cells briefly before migrating back to the apical surfaces of the cells for transmission to

    a new host. Besides transcytosis, N. meningitidis can cross the epithelium either directly

    following damage to the monolayer integrity or through phagocytes in a ‘Trojan horse’ manner.

    In susceptible individuals, once inside the blood, N. meningitidis may survive, multiply rapidly

    and disseminate throughout the body and the brain. Meningococcal passage across the brain

    vascular endothelium (or the epithelium of the choroid plexus) may then occur, resulting in

    infection of the meninges and the cerebrospinal fluid (See text for details) (Virji, M. 2009).

    The human nasopharynx is the natural biological niche colonized by N. meningitidis

    and transmission to new hosts is facilitated through aerosol droplets (Caugant, D.A.

    and Maiden, M.C. 2009), as well as direct contact. Acquisition is generally

    asymptomatic, but infrequently may result in local inflammation, invasion of mucosal

    surfaces, access to the bloodstream and fulminant sepsis or focal infections such as

    meningitis (Stephens, D.S. et al.). Meningococcal disease usually occurs 1–14 days after

    acquisition of the pathogen (Rosenstein, N.E. et al. 2001), after which the carrier state

    may be established for a period that vary between days to months. From an

    evolutionary perspective, the interactions of meningococci and the human

    nasopharynx are key events. Meningococcal carriage and transmission, not disease,

  • 10

    determine the global variation and composition of the natural population of

    meningococci.

    Colonization is an essential but considerably challenging process in meningococcal

    survival, and therefore a prerequisite for strain carriage as well as for establishing

    invasive disease (Stephens, D.S.).

    Initially, N. meningitidis preferentially adheres to relatively non-ciliated or damaged

    areas of the epithelial barrier. Pili and outer membrane opacity proteins such as Opa

    and Opc play a major role for meningococcal adhesion to human cells (Hill, D.J. et al.

    2012, Kallstrom, H. et al. 2001). Upon contact with human cells, the meningococcus

    forms microcolonies and adheres using filamentous structures named type IV pili

    (T4P) which may recognize the host receptor CD46 (Kallstrom, H. et al. 2001), forming

    a layer tightly attached to host cells (Nassif, X. et al. 1997). After this step, the capsule,

    which masks the OM proteins via steric hindrance, is lost or down-regulated due to

    cell-contact induced repression (Deghmane, A.E. et al. 2002) or selection of low or no-

    capsule expressing bacteria caused by phase variation (Hammerschmidt, S. et al. 1996).

    The absence of the capsule reveals a variety of redundant adhesins, which mediate a

    close adherence of the bacteria to host epithelial cells (Stephens, D.S. 2009). In fact,

    other minor adhesins such as Neisseria adhesin A (NadA) (Capecchi, B. et al. 2005),

    Neisseria hia/hsf homologue (NhhA) (Scarselli, M. et al. 2006), Adhesin complex

    protein (Acp) (Hung, M.C. et al. 2013), Adhesion and penetration protein (App)

    (Serruto, D. et al. 2003), Meningococcal serine protease A (MspA) (Turner, D.P. et al.

    2006) and Neisserial heparin binding antingen (NHBA) (Vacca, I. et al. 2016) have been

    shown to significantly contribute towards N. meningitidis colonization of the human

    nasopharynx.

    The interaction of bacterial opacity proteins, Opa and Opc, with CD66/CEACAMs and

    integrins respectively, on the surface of epithelial cells triggers meningococcal

    internalization (Gray-Owen, S.D. et al. 2006). Meningococci are capable of intracellular

    replication and this is in part due to iron acquisition mediated by specialized transport

    systems, such as the transferring binding protein (TbpAB), the lactoferrin binding

    protein (LbpAB), and the hemoglobin binding receptor (HmbR) (Perkins-Balding, D. et

    al. 2004). This intracellular lifestyle gives the bacteria the opportunity to evade the host

    immune response as well as to find new source of nutrients. Occasionally, bacteria can

    cross the mucosal epithelial barrier of susceptible individuals, either through

  • 11

    transcytosis or through phagocytes in a “Trojan horse” manner, or directly following

    damage to the monolayer integrity (Virji, M. 2009), and eventually enter the

    bloodstream. In healthy individuals, bacteria that cross the mucosal epithelium are

    eliminated by serum bactericidal activity. Nonetheless, survival within human blood

    relies upon different mechanisms and is dependent on their capability to evade the

    immune response and to acquire nutrients. Indeed, the up-regulation of capsule

    expression prevents antibodies and complement deposition (Uria, M.J. et al. 2008)

    hence inhibiting phagocytosis. Other strategies developed by the bacteria to evade the

    immune system are the recruitment of negative regulators of the complement cascades,

    such as Factor H (fH), which is bound by the Factor H binding protein (fHbp) (Madico,

    G. et al. 2006), or by the Neisserial surface protein A (NspA) (Lewis, L.A. et al. 2010),

    and by the Porin B (PorB) (Lewis, L.A. et al. 2013), or the recruitment of complement

    regulators, such as the C4-binding protein, which is bound by Porin A (PorA) (Jarva,

    H. et al. 2005). Once inside the bloodstream, meningococci can multiply slowly and

    eventually cross the blood-brain barrier, causing the infection of meninges and

    cerebrospinal fluid (Nassif, X. 2009). Otherwise, in case of rapid multiplication within

    the blood, the bacteria cause septicemia or meningococcemia (Rosenstein, N.E. et al.

    2001, Tinsley, C. et al. 2001).

    Overall, the onset of meningococcal disease can be seen as a failed relationship

    between the meningococcus and the host. While factors that trigger meningococcal

    entrance in the bloodstream are not yet fully understood, they are likely dependent on

    both the host and pathogen sides and include impairing of the integrity of the human

    nasopharyngeal mucosa, the lack of a protective immune response and microbial

    factors influencing virulence (Caugant, D.A. and Maiden, M.C. 2009, Stephens, D.S. et

    al. 2007).

    2.3. Meningococcal virulence factors

    Within the host N. meningitidis colonizes and invades diverse sites which represent

    different niches with respect to nutrients, environmental factors and competing

    microorganisms. The pathogen is subjected to constant selective pressures and its

    ability to rapidly adapt its metabolism and cellular composition to environmental

    changes is essential for its survival (Hill, D.J. et al. 2010). Bacteria achieve adaptation to

    the environment either by changing their genotype (genome plasticity) or by transient

  • 12

    alterations in gene expression. These two mechanisms are complementary and both

    lead to phenotypic variations.

    The genome variability is also assured by the horizontal gene transfer that occurs with

    a relatively high frequency considering the high natural competence of meningococci.

    Instead, the genome plasticity is guaranteed by the abundance of mobile elements that

    represent the 10% of the entire genome (Parkhill, J. et al. 2000). Furthermore, other

    interesting phenomenon that significantly contributes to meningococcal genome

    plasticity is phase variation. It represents an adaptive process by which N. meningitidis

    undergoes stochastic, frequent and reversible phenotypic changes as consequence of

    genetic alterations in specific loci, altering mainly virulence-associated, surface-

    exposed antigens such as outer-membrane proteins PorA, Opc, Opa, pili and specific

    adhesins, as well as LOS and capsule (Davidsen, T. et al. 2006, Feil, E.J. et al. 2001,

    Metruccio, M.M. et al. 2009, Moxon, E.R. et al. 1998). Meningococcal strains associated

    with disease have high frequency of phase variation, indicating that varying surface-

    exposed components provides substantial benefits during transmission between hosts

    (Richardson, A.R. et al. 2002).

    Distinct from phase variation, antigenic variation is a mechanism of immune evasion

    where bacteria express different moieties of functionally conserved molecules that are

    antigenically distinct within a clonal population. This process is distinct from phase

    variation, as only one variant is expressed at any given time, although the cell still

    contains the genetic information to produce a whole range of antigenic variants. In the

    pathogenic Neisseria species, antigenic variation occurs in several surface components,

    including type IV pili, LOS and Opa proteins (Davidsen, T. and Tonjum, T. 2006).

    The virulence of N. meningitidis is influenced by multiple factors that are mainly

    located in the outer membrane (Figure 2.3).

  • 13

    Figure 1.3 Meningococcal cell compartments. Schematic representation of the different

    bacterial compartments and of the main components of the outer membrane, together with their

    known function (adapted from Rosenstein NE, 2001).

    The main virulence factor is the polysaccharidic capsule, which represents a barrier

    that protects the bacterium from the host innate and adaptive immune system

    (Schneider, Exley, Ram, Sim, & Tang, 2007; Vogel & Frosch, 1999). It also defends

    meningococcus from desiccation during airborne transmission between hosts (Virji, M.

    2009); (Romero, J.D. et al. 1997). Its expression is phase variable (Hammerschmidt, S. et

    al. 1996) and the switching of the capsule locus between strains confers a selective

    advantage to the bacterium for its evasion to opsonization or neutralization by natural

    or vaccine-induced anti-capsular antibodies (Swartley, J.S. et al. 1997). In

    meningococcus, LPS are referred to as lipooligosaccharide (LOS) because of the

    presence of repeating short saccharides instead of long-chain saccharides. LOS is the

    major constituent of the outer leaflet of the meningococcal outer membrane (OM),

    responsible for the physical integrity and proper functioning of the membrane and

    required for resistance of N. meningitidis to complement (Geoffroy, M.C. et al. 2003).

    LOS comprises an inner and outer oligosaccharide core attached to the lipid A portion

    that anchors the LOS in the outer leaflet of the OM. Lipid A is responsible for the

    toxicity of LOS due to its ability to bind to different Toll-like receptors on monocytes

    and on dendritic cells triggering the secretion of various inflammation mediators

    (Brandtzaeg, P. et al. 2001); (Braun, J.M. et al. 2002). Phase and antigenic variations lead

    to different saccharide chains altering dramatically the antigenic properties of LOS and

    enabling individual meningococci to display a repertoire of multiple LOS structures

  • 14

    simultaneously (Jennings, M.P. et al. 1999). Another group of virulence factors

    involved in the interface between the meningococcus and the host are pili. They are

    long filamentous structures consisting of protein subunits that extend from the

    bacterial surface beyond the capsule (Pinner, R.W. et al. 1991, Virji, M. et al. 1992). Pili

    represent the major contributor to the adhesive property of the capsule (Stephens, D.S.

    et al. 1981, Virji, M. et al. 1991) and in addition they are involved in the uptake of

    foreign DNA from the extracellular environment, hence increasing transformation

    frequency and consequently genetic adaptability (Helaine, S. et al. 2007).

    Furthermore, the presence of other OM-associated proteins is important in host cell

    interaction. The opacity proteins (Opa and Opc) are integral outer membrane proteins

    that mediate pathogen-host interaction, adhering to and invading of epithelial and

    endothelial cells (Virji, M. et al. 1992). A key role in the adhesion is carried out by

    adhesins, which are generally low expressed in vitro, but they might be upregulated in

    vivo. In fact, they may undergo to antigenic variation and/or phase variation, hence

    allowing the meningococcus to evade the immune system and adapt to different niches

    (Virji, M. 2009). The Neisserial adhesin A (NadA) is a surface-exposed member of the

    Oligomeric coiled-coil adhesin family of bacterial Trimeric Autotransporter adhesins

    (El Tahir, Y. et al. 2001, Helaine, S. et al. 2007). NadA mediates adhesion to and

    invasion of human epithelial cells (Capecchi, B. et al. 2005), suggesting its pivotal role

    in the adhesion to the naso- and oro-pharyngeal epithelia during meningococcal

    colonization of the human upper respiratory tract. Other adhesins have been reported

    to play a role in colonization and/or invasion. NHBA has been recently shown to

    participate during the colonization process by increasing adherence to host tissues by

    binding glycosaminoglycans (Vacca, I. et al. 2016), and mediating biofilm formation

    ((Arenas, J. et al. 2013)). The Meningococcal surface fibril (Msf), previously termed

    Neisseria hia/hsf homologue A (NhhA) (Peak, I.R. et al. 2000, Weynants, V.E. et al.

    2007), mediates adhesion to epithelial cells and to components of the extracellular

    matrix, even though at low levels (Scarselli, M. et al. 2006). Moreover, it has been

    shown its involvement in the immune system evasion. Msf binds to the activated form

    of Vitronectin and inhibits the terminal complement pathway (Griffiths, N.J. et al.

    2011), and its role in inhibiting phagocytosis, inducing macrophages apoptosis and

    protecting bacteria against complement-mediated killing has been suggested

    (Sjolinder, H. et al. 2008, Sjolinder, M. et al. 2012). Two homologous autotransporters,

  • 15

    the Adhesion penetration protein (App) and the Meningococcal serine protease A

    (MspA) are involved in the bacterial interaction to epithelial cells (Serruto, D. et al.

    2003, Turner, D.P. et al. 2006) and also in the apoptosis of dendritic cells (Khairalla, A.S.

    et al. 2015). Glycolipid adhesins such as members of the Multiple adhesin family (Maf)

    may contribute to the bacterial interaction with host cells (van Putten, J.P. et al. 1998).

    Interestingly they are found to be associated with genomic islands present only in

    pathogenic Neisseria species, both meningococcus and gonococcus (Jamet, A. et al.

    2015).

    The two porins PorA and PorB, are the most abundant proteins present in the

    meningococcal OM. They are composed of relatively conserved regions, which are

    predicted to form the β-barrel structure that spans through the membrane, alternated

    with variable regions, which should be surface-exposed, hence undergoing to a strong

    selective pressure. The formation of trimers creates the pore structure that allows the

    passage of small hydrophilic solutes necessary for the bacterial metabolism. Porins

    were shown to be interacting with several human cell types and proteins (Orihuela,

    C.J. et al. 2009); moreover, PorA elicits a protective immune response in humans

    (Holst, J. et al. 2009, Wedege, E. et al. 1998), while PorB might be involved in the

    immune system evasion by binding the human fH (hfH) (Lewis, L.A. et al. 2013). The

    regions of PorA that generate the immune response are loops 1 and 4, named VR1 and

    VR2, that are hyper variable among strains. OMV based vaccines, such as 4CMenB, use

    PorA as significant antigen generating bactericidal immune responses. However, due

    to the hyper variability of the immune-dominant regions, PorA-based vaccines provide

    protection only against strains expressing homologous PorA serosubtypes (see below).

    Furthermore, the genome of N. meningitidis contains a set of membrane-associated

    factors responsible for the host’s immune system evasion and hence for its virulence.

    As indicated by the elevated susceptibility to microbial, including meningococcal,

    infections exhibited by individuals with complement deficiencies (Figueroa, J. et al.

    1993). In order to escape from the innate immune system, N. meningitidis has evolved a

    plethora of mechanisms that target the complement cascades. As already introduced

    above, at least three meningococcal proteins have shown to bind the fH, fHbp (Madico,

    G. et al. 2006), NspA (Lewis, L.A. et al. 2010) and PorB (Lewis, L.A. et al. 2013). Strains

    lacking both fHbp and NspA were not able to bind fH and indeed were more

    susceptible to complement-dependent killing (Echenique-Rivera, H. et al. 2011, Lewis,

  • 16

    L.A. et al. 2010). In addition, the observed binding of heparin from the NHBA may

    increase bacterial serum resistance due to the potential interactions of heparin with fH

    (Serruto, D. et al. 2010).

  • 17

    2.4. Anti-meningococcal vaccines

    Due to its rapid progression and the difficulties to diagnose it (Rosenstein, N.E. et al.

    2001, Thompson, M.J. et al. 2006), the most effective option to prevent meningococcal

    disease is vaccination. No broadly protective vaccine is currently available to provide

    protection against all serogroups of N. meningitidis. Different meningococcal vaccines

    have been developed against the distinct serogroups (Zahlanie, Y.C. et al. 2014). There

    are a number of polysaccharide and conjugate meningococcal vaccines in use against

    serogroups A, C, Y and W135. The tetravalent vaccine composed of purified capsular

    polysaccharides, although efficacious in adolescent and adults, is poorly immunogenic

    in infants and fails to induce immunological memory. However, when conjugated to a

    carrier protein, capsule polysaccharides show a greatly improved immunogenicity in

    young infants (Granoff, D.M. et al. 2007, Nassif, X. 2009, Virji, M. 2009). Monovalent,

    bivalent, and tetravalent polysaccharide conjugative vaccines are available and

    effective against meningococcal serogroups A, C, Y and W-135 (Zahlanie, Y.C. et al.

    2014); http://www.who.int/ith/vaccines/meningococcal/en/]. The first trials

    conducted in the United Kingdom with the meningococcus C conjugate showed a

    dramatic decline in the incidence of serogroup C disease in all age groups that received

    the vaccine (Borrow, R. et al. 2000, Miller, E. et al. 2001) with an efficacy of 97 and 92

    per cent for teenagers and toddlers, respectively (Ramsay, M.E. et al. 2001).

    In contrast, the group B capsule polysaccharide is not suitable as vaccine antigen. It

    consists of a homolinear polymer of α(2→8)N-acetyl neuraminic acid, also known as

    polysialic acid, which is structurally similar to the sialic acid found in human neural

    tissue, hence is poorly immunogenic in humans and may elicit auto-antibodies (Finne,

    J. et al. 1987, Finne, J. et al. 1983). Therefore, efforts to develop a vaccine against

    meningococcus serogroup B (MenB) focused mainly on non-capsular antigens, such as

    proteins or LOS. The principal challenge has been to identify surface-exposed non-

    capsular antigens that are safe, antigenically conserved and that elicit a broad Serum

    Bactericidal Antibody (SBA) response. Licensed and promising group B vaccine

    approaches are discussed below.

    http://www.who.int/ith/vaccines/meningococcal/en/

  • 18

    2.4.1. Licensed vaccines against MenB

    Detergent-extracted OMV vaccines (dOMV)

    In order to control outbreaks caused by specific MenB strains vaccines composed of

    dOMV have been successfully employed in Norway (Fredriksen, J.H. et al. 1991), Cuba

    (Sierra, G.V. et al. 1991), Chile (Boslego, J. et al. 1995) and New Zealand (Oster, P. et al.

    2005). The detergent treatment removes the toxic LOS, but it also extracts other

    desirable antigens such as lipoproteins. Consequently, the porin protein PorA results

    to be the immuno-dominant antigen (Martin, D.R. et al. 2006, Tappero, J.W. et al. 1999).

    Despite dOMV vaccines resulted to be safe and to induce good functional responses in

    humans, the immune response elicited is effective only against strains expressing the

    same PorA serosubtype, due to PorA antigenic variability (van der Ley, P. et al. 1991).

    Therefore, dOMV vaccines are well-suited to control local, clonal outbreaks but they do

    not confer broad protection.

    4CMenB

    The advent of the genomic era and the availability of whole genome sequences have

    contributed to radically change the approach to vaccine development. Indeed, the in

    silico approach named Reverse Vaccinology (RV) aims to identify surface-exposed

    non-capsular antigens that are antigenically conserved among strains and elicit a

    bactericidal serum response. This approach led to the development of the four

    component recombinant protein vaccine 4CMenB (Giuliani, M.M. et al. 2006, Giuliani,

    M.M. et al. 2010). 4CMenB contains five Genome-derived Neisseria Antigens (GNA)

    formulated together with the dOMV component from the NZ98/254 strain (Martin,

    D.R. et al. 2006). Based on their ability to induce broad protection three major antigens

    have been selected (Giuliani, M.M. et al. 2006): NadA (Capecchi, B. et al. 2005,

    Comanducci, M. et al. 2002) is present as single polypeptide, while fHbp (Beernink,

    P.T. et al. 2008, Masignani, V. et al. 2003) and NHBA (Serruto, D. et al. 2010, Welsch,

    J.A. et al. 2003) are fused to the conserved meningococcal gene products GNA2091 and

    GNA1030, respectively. The other two antigens, GNA2091 and GNA1030, are well

    conserved in N. meningitidis, but less functionally characterized than the other antigens

    (Bos, M.P. et al. 2014, Donnarumma, D. et al. 2015, Muzzi, A. et al. 2013). They were

  • 19

    included in the vaccine formulation since they increase immune responses to the main

    vaccine antigens when present as fusion proteins with the respect of the individual

    antigens (Giuliani, M.M. et al. 2006). 4CMenB was licensed in Europe in 2013 and in the

    U.S. in 2015, following its progression through clinical trials that have demonstrated its

    safety (Esposito, S. et al. 2014, Prymula, R. et al. 2014, Toneatto, D. et al. 2011) and its

    efficacy in inducing a protective immune response in infants, children, adolescents and

    adults against the majority of MenB strains (Gossger, N. et al. 2012, Kimura, A. et al.

    2011, McQuaid, F. et al. 2014, Read, R.C. et al. 2014, Santolaya, M.E. et al. 2012, Snape,

    M.D. et al. 2013, Vesikari, T. et al. 2013).

    Figure 2.4 Schematic representation of the 4CMenB vaccine antigens on the surface

    of N. meningitidis (from Serruto D, 2012). The different bacterial compartments (outer

    membrane, periplasmic space, cytoplasmic membrane) and the main antigens

    identified through reverse vaccinology approach (NHBA, fHbp and NadA) are

    depicted. Other components of the meningococcal membranes are also shown (pilus,

    polysaccharide capsule, lipooligosaccharide and integral inner and outer membrane

    proteins).

    Bivalent fHbp-based vaccine or Trumenba

    Trumenba was licensed in the U.S. in 2014 for a target population of adolescents and

    young adults. However, it is not suitable for use in infants considering that it consists

    of purified lipoproteins known as TLR-2 agonists (Richmond, P.C. et al. 2012). It is a

    recombinant protein-based vaccine composed of equal amounts of two variants,

  • 20

    subfamily A05/var3.45 and subfamily B01/var1.55, of lipidated fHbp (Fletcher, L.D. et

    al. 2004).

    2.4.2. Investigational MenB vaccines

    Recombinant protein vaccines

    Several protein antigens have been investigated for their protective ability for use in a

    MenB vaccine, among which NspA, TbpB, FetA, ZnuD and others (Halperin, S.A. et al.

    2007, Hubert, K. et al. 2013, West, D. et al. 2001). The main issue with all of these

    approaches was the limited cross protective potential of any one antigen. It was clear

    that a multivalent approach was needed to guarantee a wide protection.

    Native Outer Membrane Vesicles (nOMV) vaccines

    nOMV are spherical portions of the OM, ~20–250 nm in diameter, produced by Gram

    negative bacteria. They are spontaneously released during the active growth into the

    surrounding medium. These portions of the OM bud and detach from the cell,

    enclosing many native bacterial antigens and periplasmic constituents (Figure 2.5).

    The vesicles play diverse roles like delivery of virulence factors , modulation of the

    host immune system during pathogenesis, aid in nutrient acquisition, mediation of

    cellular communication, surface modifications and the elimination of undesired

    components that, ultimately, make them a transportable part of the bacterial arsenal

    and survival system (Collins, B.S. 2011, Kuehn, M.J. et al. 2005, Schwechheimer, C. et

    al. 2015).

  • 21

    Figure 2.5 Model of native Outer Membrane Vesicles (nOMV) biogenesis. NOMV vesicles are

    proteoliposomes consisting of OM phospholipids and LPS, a subset of OM proteins and

    periplasmic (luminal) proteins (Kuehn, M.J. and Kesty, N.C. 2005).

    nOMV represent an attractive vaccine platform mimicking the bacterial cell surface.

    Since nOMV do not undergo to a detergent extraction procedure, like dOMV do, they

    preserve high amounts of lipooligosaccharide (LOS) as well as protective lipoproteins

    which would otherwise be removed by the detergent. This was expected to improve

    immunogenicity and cross-protection provided but it raised safety concerns.

    Consequently, to prepare safe nOMV vaccines the strain must be genetically

    engineered to reduce the LOS reactogenicity. The acylation of lipid A molecule is

    responsible for its endotoxin activity and two mutations (lpxL1 and lpxL2), affecting

    the reduction of lipid A, have been successfully exploited (Bonvehi, P. et al. 2010,

    Keiser, P.B. et al. 2011, Keiser, P.B. et al. 2010, Koeberling, O. et al. 2011). The lpxL1

    gene, homologous to E.coli htrB, encodes for a late acyltransferase of lipid A

    biosynthesis. Its deletion lead to penta- instead hexa-acetylated molecules, resulting in

    lower endotoxin activity of LOS (van der Ley, P. et al. 2001). Instead the lpxL2 gene,

    homologous of E.coli lpxLM, encodes for a lauroyl acyltransferase. Its deletion leads to

    a tetra-acylated lipid A lacking both secondary lauroyl chains.

    nOMV vaccines prepared from wild-type strain were poorly immunogenic in mice

    (Koeberling, O. et al. 2008, Moe, G.R. et al. 2002). Koeberling and colleagues

  • 22

    demonstrated that the level of expression of a key antigen, as fHbp, was a critical

    parameter to elicit broad serum bactericidal responses (Koeberling, O. et al. 2011). The

    overexpression of some antigens (Keiser, P.B. et al. 2011) and the removal of the

    immunodominant PorA antigen (Bonvehi, P. et al. 2010) were two strategies tested in a

    phase I clinical trial. In the first case, the nOMV vaccine resulted to be safe and

    immunogenic (Keiser, P.B. et al. 2011); nevertheless, the major contribution to

    bactericidal activities was from antibodies raised from LOS providing immunotype

    specific bactericidal responses. This result was suggested to be due to insufficient

    levels of the antigens over-expressed on the vesicles (Koeberling, O. et al. 2011). In the

    second case, PorA was deleted to avoid its immune-dominance. The prototype vaccine

    strain was also engineered to express a truncated form of LOS immunotype L3,7 that is

    the most common in invasive MenB strain (Scholten, R.J. et al. 1994). This OMV-based

    vaccine offered good safety but low immunogenicity in healthy young adults (Bonvehi,

    P. et al. 2010, Weynants, V. et al. 2009).

  • 23

    Chapter 1

    NHBA regulation and expression during colonization and invasion

  • 24

    Sensing the environment

    Colonization is an essential as well as a considerably challenging process in

    meningococcal survival, and therefore a prerequisite for strain carriage as well as for

    establishing invasive disease (Stephens, D.S. 2009). The nasopharyngeal epithelium is a

    complex ecological niche that poses several hurdles for bacterial colonization and

    survival. Compounds such as mucus, antimicrobial peptides and immunoglobulins

    provide physical and biochemical host defenses (Laver, J.R. et al. 2015). Furthermore,

    this environment is deprived of nutrients such as iron, carbohydrates and oxygen

    essential for bacterial growth and N. meningitidis therefore needs to compete for these

    limited nutrients with the resident microflora. Taken together, these factors make de

    novo colonization and survival challenging. N. meningitidis is incredibly well adapted

    to this environment and has developed several mechanisms to control expression of

    adhesion molecules (Deghmane, A.E. et al. 2002) (Grifantini, R. et al. 2002, Hey, A. et al.

    2013), biofilm formation (Arenas, J. et al. 2013), iron acquisition (Larson, J.A. et al. 2002,

    Schryvers, A.B. et al. 1999), metabolism (Jamet, A. et al. 2009, Mendum, T.A. et al. 2011)

    (Laver, J.R. et al. 2015), and immune evasion factors (Lomholt, H. et al. 1992,

    Yazdankhah, S.P. et al. 2004) .

    One of the key signals sensed by N. meningitidis to determine its environment and to

    induce the expression of either adhesion or immune evasion factors is temperature

    (Laver, J.R. et al. 2015). Temperatures within the upper respiratory tract are affected by

    the passage of air during respiration of the host, the precise anatomical location and

    the presence of local inflammation (Keck, T. et al. 2000, McFadden, E.R., Jr. et al. 1985)

    (Figure 2.6). These factors can result in an overall variability of temperature in this

    niche ranging from 25.3±2.1°C in the nasal vestibule to 33.9±1.5°C in the nasopharynx,

    generally being several degrees below core body temperature (Keck, T. et al. 2000). N.

    meningitidis has evolved to rapidly and efficiently adapt its metabolism to even minor

    temperature changes in the environment. During the development of invasive disease,

    N. meningitidis passes from the lower temperatures in the upper airway to the core

    body temperature of 37°C or higher with a febrile response to infection (Cabanac M.

    1990). Within the bloodstream although the increased temperature and the abundance

    of nutrients promote the fast growth of the bacterium, the presence of the complement

    cascade components, immunoglobulins and immune cells represent a big threat,

    meanwhile. Therefore a rapid adaptation to the new environment is required, in fact

  • 25

    approximately 30% of the genes in the genome are dramatically regulated on entry to

    whole human blood (Echenique-Rivera, H. et al. 2011), triggering an immune evasion

    response. Key antigens and virulence factors such as capsule biosynthesis (CssA),

    sialylation of LPS (Lst) and fHbp involved in immune evasion and in establishing

    invasive disease, show increased expression at 37°C relative to lower temperatures

    (Loh, E. et al. 2013, Loh, E. et al. 2016). However, the role of lower temperature on the

    expression of virulence factors has received considerably less attention. Recently, a

    comparative proteomic study showed that 375 proteins were differentially expressed

    between 32°C and 37°C (Lappann, M. et al. 2016).

    Figure 2.6 Within the host N. meningitidis encounters different niches. Temperature is one of

    the key signal sensed by N. meningitidis to determine its environment. The temperatures that N.

    meningitidis encounters during transmission, colonization and invasion are reported.

    Neisserial Heparing Binding Antigen (NHBA)

    NHBA is a surface exposed lipoprotein that is specific to Neisseria species. NHBA is

    one of the major antigens of the serogroup B meningococcal vaccine, 4CMenB (Serruto,

    D. et al. 2012), and induces antigen-specific bactericidal antibodies in both animals and

    humans (Serruto, D. et al. 2010); (Giuliani, M.M. et al. 2010). The nhba gene is

  • 26

    ubiquitous in meningococcal strains of all serogroups and it is also found in N.

    gonorrhoeae as well as in different commensal neisserial species (Bambini, S. et al. 2009);

    (Jacobsson, S. et al. 2006, Muzzi, A. et al. 2013). Analysis of gene sequences from

    genetically diverse serogroup B strains revealed the existence of more than 400 distinct

    peptides, which are associated with clonal complexes and sequence types

    (Comanducci, M. et al. 2002, Jacobsson, S. et al. 2006, Muzzi, A. et al. 2013).

    Considerable variation is observed at the level of primary amino acid sequence which

    ranges in length from approximately 430 to 500 residues (Figure 2.7). Most variability

    is observed at the level of the amino-terminal region, which is annotated as

    intrinsically unfolded by commonly used structure prediction algorithms (Vacca, I.

    2014). In contrast, the carboxyl-terminal region consists of a single 8-stranded anti-

    parallel beta-barrel structure and is highly conserved (Esposito, V. et al. 2011).

    The two domains are linked through an arginine-rich motif which is responsible for

    NHBA binding to heparin in vitro and contributes to increased survival of the un-

    capsulated N. meningitidis in human serum (Esposito, V. et al. 2011, Serruto, D. et al.

    2010). Conversely, it has been recently shown that NHBA plays an integral part in

    binding heparin sulfate proteoglycans on epithelial cells and thus directly mediates

    adhesion of N. meningitidis (Vacca, I. et al. 2016). Furthermore, the presence of this

    arginine-rich domain was implicated in DNA-binding and could therefore also play a

    role in the formation of neisserial microcolonies and biofilms (Arenas, J. et al. 2013).

    NHBA can be processed by the meningococcal protease NalP and human lactoferrin

    (hLf). Cleavage occurs either upstream and downstream of the NHBA Arg-rich region

    resulting in one of two possible cleavage fragments termed C2 and C1, respectively

    (Serruto, D. et al. 2010).

    It was also demonstrated that the C-terminal fragment (C2), released upon NalP

    proteolysis, alters endothelial cell permeability by inducing the internalization of the

    adherens junction protein VE-cadherin, which is in turn responsible for the endothelial

    leakage. Thus, the NHBA-derived fragment C2 might contribute to the extensive

    vascular leakage typically associated with meningococcal sepsis (Casellato, A. et al.

    2014).

  • 27

    Figure 2.7 NHBA protein schematic view and variability. NHBA protein sequence reflects a

    modular structural organization, where it is possible to recognize three main domains (A, B and

    C). The presence of an insertion sequence of 60 amino acids, present only in some of the NHBA

    peptides (Insertion IB), allows to discriminate between long or short isoforms. Functional sites

    are represented by the Arg-rich region (in brown), by the NalP cleavage site (in green) and by

    the human lactoferrin cleavage site (in grey). The C-term of the protein, corresponding to

    module C is highly conserved and is represented by a beta-barrel structure. The lower graph

    shows the percentage of amino acid conservation between the different peptides along the

    protein sequence (adapted from (Vacca, I. 2014)).

    The upstream regulatory region of the nhbA gene is characterized by the presence of

    the 150-bp Contact Regulatory Element of Neisseria (CREN) in strains, such as MC58,

    belonging to clonal complex ST-32. This regulatory element is specific to pathogenic

    Neisseria species and is involved in the induction of the downstream associated genes

    upon contact with target eukaryotic cells (Deghmane, A.E. et al. 2002). NHBA

    expression is known to be induced after incubation of bacteria with epithelial cells in

    the CREN-containing strain MC58, while its expression remains unaltered in the

    CREN-lacking strain 8013 (Deghmane, A.E. et al. 2003). It was therefore proposed that

    cell contact increased NHBA levels on meningococcal surface in the ST-32 invasive

    hypervirulent strains and that increased expression of nhba upon host contact might at

    least partially account for the hypervirulent phenotype of this clonal complex.

  • 28

    3. Results

    3.1. NHBA expression and surface exposure are temperature-dependent

    NHBA is an important virulence factor for N. meningitidis and it is also protective

    antigen able to elicit an immune response in preclinical and clinical trials (Serruto, D. et

    al. 2010). Therefore, understanding the mechanisms that drive NHBA regulation is an

    important goal to better understand N. meningitidis pathogenesis and vaccine induced

    response. We therefore investigated how physiologically relevant temperatures, which

    mimic the different stages of pathogenesis, may affect the expression levels of NHBA.

    Strains MC58, M11719 and 8047 were grown overnight on GC agar plates at

    physiologically relevant temperatures ranging from 28°C up to 40°C. We found that

    NHBA expression was thermoregulated in an inverse manner to fHbp, with higher

    expression at lower temperature. Western blot analysis showed that NHBA expression

    was at its highest level between 28°C and 30°C in all these strains and that its

    expression decreased markedly with increasing temperatures (Figure 3.1 A). In

    contrast, fHbp expression was highest at elevated temperatures and decreased with

    temperature reduction. In order to understand whether temperature regulation of

    NHBA was conserved among different N. meningitidis isolates, we expanded our

    analysis to a broader panel of strains belonging to different clonal complexes, carrying

    different variants and also long or short isoforms of NHBA (Figure 3.1 B and Table 3.1).

    NHBA expression levels were variable among the different strains and showed

    different processing patterns depending on the strain background and NHBA

    variant/isoform present (Table 3.1). Despite different variants and expression levels

    between the strains tested, all strains showed increased levels of NHBA at 30°C

    compared to 37°C. As NHBA is a surface exposed neisserial protein, we confirmed that

    increased expression levels of NHBA also resulted in increased levels of NHBA

    exposed on the bacterial cell surface using flow cytometry (Figure 3.1 C).

  • 29

    Figure 3.1 NHBA expression and surface exposure are increased at reduced temperatures. (A)

    The defined strains were grown overnight on GC agar plates at the indicated temperatures.

    Whole cell lysates were prepared and separated by SDS-PAGE prior to Western Blotting. The

    indicated proteins were detected using mouse-polyclonal antisera. Hfq served as loading

    control between different samples. In MC58 strain the full-length protein migrates with an

    apparent molecular weight of approximately 62 kDa, while other bands at approximately 49

    kDa results from the bacterial proteases’s processing (Serruto, D. et al. 2010). (B) The defined

    strains were grown in GC broth at 30°C or 37°C until OD600 0.25. Whole cell lysates were

    prepared and separated by SDS-PAGE prior to Western Blotting. Different strains express

    different variants and isoforms of NHBA (See table 3.1 for strains details), however the bands

    specificity is confirmed by the nhba deletion mutant (Δnhba) generated for each strain. (C) Flow

    cytometric analysis of strain MC58 showing surface exposure of NHBA at the indicated

    temperatures.

  • 30

    Strain Country of origin

    Year of isolation

    Capsular group

    Clonal complex

    NHBA peptide variant

    NHBA isoform

    M11205 USA 2003 B 41/44 p0001 Long

    M11822 USA 2004 B 41/44 p0001 Long

    NGH38 Norway 1988 B uaa p0002 Long

    MC58 UK 1985 B 32 p0003 Long

    M10935 USA 2003 B 35 p0058 Long

    M14933 USA 2006 B 32 p0003 Long

    M10713 USA 2003 B 41/44 p0010 Short

    M03279 USA 1997 B 41/44 p0011 Short

    N16/07 Norway 2007 B 41/44 p0029 Long

    M11204 USA 2003 B 41/44 p0029 Long

    M10282 USA 2003 B 41/44 p0002 Long

    M07-0240679 UK 2007 B 269 p0017 Short

    M11719 USA 2003 B 162 p0020 Short

    M16453 USA 2007 B 41/44 p0144 Short

    M18070 USA 2008 B 162 p0020 Short

    8047 USA 1978 B 11 p0020 Short

    Table 3.1 List of natural strains reported in Figure 3.1. Main characteristics are indicated. For

    each of them a nhba deletion mutant was generated. *Unassigned

  • 31

    3.2. Mutations and deletions in the 5’UTR and 5’TR of nhba affect expression

    The nhba gene was originally annotated as NMB2132 according to its location within

    the genome sequence of the strain MC58 (Tettelin, H. et al. 2000). MC58 nhba locus

    schematic view (upper panel) and details of the intergenic region and the 5’TR of nhba

    (lower panel) are reported in Figure 3.2 A. The conservation of this region among all

    the Neisseria species present in the PubMLST database is also reported (mid panel). The

    region shows a very good conservation among 8373 Neisseria strains (green and

    greeny-brown bars). Red bars indicate the lack of conservation (5.8% strains) which

    corresponds to the CREN sequence, specific for ST-32.

    Upstream and in the same orientation of nhba is located NMB2133. A Rho-independent

    terminator is predicted around 22 nucleotides downstream (Figure 3.2 A lower panel).

    qRTPCR experiments confirmed that no co-transcription is detectable among NMB2133

    and NMB2132 (data not shown). A putative promoter (Pnhba) was identified upstream

    the CREN sequence.

    The annotated translational start site is boxed in black and the ribosomal binding site is

    also indicated and underlined. However, in frame with the annotated one and just next

    to it, two more putative translational start sites were identified (boxed with dashed

    lines). Moreover, within the CREN sequence and still in frame with the annotated one,

    another putative translational start site (boxed in green) was identified, carrying also

    an alternative ribosomal binding site (underlined in green) (Deghmane, A.E. et al.

    2003). In order to investigate which one corresponds to the initiation of translation in

    correlation with temperature changes, a series of site-directed mutagenesis were

    performed (Figure 3.2 B). As shown by Western blot analysis, small deletions or single

    base mutations affecting these sites (Mut_1-4) led to decreased or abolished NHBA

    expression, without affecting thermoregulation (Figure 3.2 C left panel). We identified

    a T-rich region in the 5’TR of nhba, 20 nucleotides downstream to the putative

    translational start site. In silico secondary structure prediction of 5’UTR+50bp in the

    coding sequence suggested a direct interaction between the T-rich region and the

    ribosomal binding site (data not shown). Synonymous mutations in this region (Mut_5-

    7) led to an overall decreased expression of NHBA, without affecting NHBA

    thermoregulation (Figure 3.2 C right panel).

  • 32

    A

    B

    C

  • 33

    Figure 3.2 Schematic representation of nhba locus in MC58 strain and site-directed

    mutagenesis. (A) Schematic representation of the nhba locus (upper panel). The conservation of

    the indicated intergenic region obtained by multiple sequence alignments of 8373 Neisseria

    strains present in PubMLST database is shown (mid panel). Each bar represents the percentage

    of conservation of the corresponding nucleotide. Green bar = 100% identity; Greeny-brown bar

    < 100% identity; Red bar < 30% identity. The nucleotide sequence of the indicated region is

    reported (lower panel): the 3’ region of NMB2133 is boxed in dark grey and the 5’ region of nhba

    (NMB2132) is boxed in light grey. The nucleotides pairing in the stem region of the Rho-

    independent terminator are underlined (dot line). In the intergenic region downstream of

    NMB2133, a putative promoter sequence was identified (Pnhba). The -35 and -10 elements of the

    Pnhba are indicated and the putative transcriptional start site is indicated and highlighted in

    bold. The contact regulatory element of Neisseria (CREN), a 150-bp sequence specific for

    pathogenic Neisseria species, is present in MC58 strain immediately upstream of the ribosomal-

    binding site (Deghmane, A.E. et al. 2003) and is boxed in green. The ribosome binding sites are

    underlined and the translation start sites are boxed. (B) Schematic representation of site-

    directed mutagenesis. Red dashes indicate nucleotides deletion, red nucleotides indicate non-

    synonymous mutations (Mut_3 and Mut_4) and synonymous mutations (Mut_5-Mut_7). (C)

    Mutant strains were grown overnight on GC agar plates at the indicated temperatures. Whole

    cell lysates were prepared and separated by SDS-PAGE prior to Western Blotting. The indicated

    proteins were detected using mouse-polyclonal antisera. The *a symbol indicates a non-specific

    band used as loading control between different samples.

  • 34

    3.3. NHBA is expressed during the active growth

    Previous analysis only represented a single time point of NHBA expression levels

    during mid-exponential growth. In order to determine how NHBA expression

    progresses during the entire growth of N. meningitidis, we grew NGH38 strain at either

    30°C or 37°C in 50 ml of liquid culture. We took samples for RNA extraction and

    Western Blotting at various stages during the entire growth curve of the strain (Figure

    3.3). By culturing the strain in flasks, we were able to take samples every hour and

    follow the growth for ten hours (Figure 3.3 A). Both curves reached high OD600 values

    however, under these experimental conditions, bacteria grown at 30°C showed longer

    lag phase (T0-T4) and slightly lower OD600 values at the end of the growth, respect to

    those grown at 37°C. Firstly, we investigated the transcriptional profile of nhba, fHbp

    and adk during growth in liquid culture (Figure 3.3 B). We determined by qRT-PCR

    that nhba transcript was most abundant during active bacterial replication. Once

    bacteria entered stationary phase, transcription of nhba was almost abolished. During

    the active replication, nhba RNA steady state levels were higher at 30°C respect to 37°C.

    Conversely, fhbp transcript resulted to be slightly more abundant at 37°C respect to

    30°C, while no differences were observed for adk comparing the two temperatures. The

    nhba RNA steady state level expression profiles during the entire growth were

    confirmed also at protein level by Western blotting, showing that NHBA is most

    abundant during the active growth at both temperatures (Figure 3.3 C).The higher

    expression of NHBA also resulted in higher processing at 30°C.

  • 35

  • 36

    Figure 3.3 NGH38 growth profiles and NHBA expression levels during the entire growth. (A)

    Growth profiles of NGH38 strain in 50 ml of MCDMI liquid medium at 30°C (blu line) or 37°C

    (red line). Bacteria were grown for 10 hours and RNA isolation and samples for Western blot

    analysis were collected every hour. (B) nhba, fHbp and adk RNA steady state levels were

    quantified by qRT-PCR and relative expression levels were determined normalizing to 16S-

    rRNA. (C) Western blot analysis of NHBA on whole cell lysates collected at the indicated time

    points. In NGH38 strain the full-length protein migrate with an apparent molecular weight of

    approximately 62 kDa, while other bands at approximately 49 kDa results from the bacterial

    proteases’s processing (Serruto, D. et al. 2010). The *a symbol indicates a non-specific band used

    as loading control.

    Therefore we decided to compare more in details the expression of NHBA in response

    to temperature by using standard in vitro conditions. We grew MC58 strain at either

    30°C or 37°C in 7 ml liquid culture and took samples for RNA extraction and Western

    Blotting at various stages during the entire growth curve of the strain (Figure 3.4 A).

    Firstly, we investigated the transcriptional profile of nhba during growth in liquid

    culture (Figure 3.4 B). By qRT-PCR we confirmed that nhba transcript was most

    abundant during active bacterial replication. Once bacteria entered stationary phase,

    transcription of nhba was almost abolished (Figure 3.4 B). Although there was a trend

    for slightly increased nhba transcript levels at 30°C, these transcriptional differences

    were only significant in late exponential phase. We therefore next examined the NHBA

    protein expression profile at the same growth phase as the nhba transcript levels. In

    accordance with the data for the nhba transcript, we observed that NHBA level at each

    temperature were highest during exponential growth of the bacterium (Figure 3.4 C).

    As seen previously, NHBA protein levels in each growth phase were always higher at

    30°C relative to 37°C (Figure 3.4 C). However, although nhba transcript is barely

    detectable at stationary phase with no differences between the two temperatures,

    becomes evident that the protein is still abundant in bacteria cultured at 30°C. To

    quantify the differences in protein expression levels between the two temperatures

    tested, we used relative protein quantification (Figure 3.4 D). This analysis determined

    that the amount of NHBA at 30°C was approximately 3-5-fold higher than at 37°C.

  • 37

    Figure 3.4 NHBA is expressed during the exponential phase and its expression at 30°C is

    higher relative to 37°C at both RNA and protein levels. (A) Growth profiles of MC58 strain in

    GC liquid medium at 30°C (continuous line) or 37°C (dot line). Samples for western blot

    analysis and RNA isolation were collected at early exponential phase (OD600 ̴0.3), late

    exponential phase (OD600 ̴0.9) and at stationary phase (OD600 ̴1.1), as indicated by the star

    symbols. (B) nhba RNA steady state levels were quantified by qRT-PCR and relative expression

    levels were determined normalizing to 16S-rRNA. (C) Western blot analysis of NHBA on whole

    cell lysates collected at the indicated time points. (D) Relative protein quantification performed

    with ImageJ 1.6 software. In MC58 strain the full-length protein migrate with an apparent

    molecular weight of approximately 62 kDa, while other bands at approximately 49 kDa results

    from the bacterial proteases’s processing (Serruto, D. et al. 2010). The *a symbol indicates a non-

    specific band used as loading control and for relative protein quantification. All the data

    represent the mean +/- SEM from three independent biological replicates and were analyzed by

    Two-way Anova followed by uncorrected Fisher’s LSD multiple comparison test (**** p

  • 38

    3.4. NHBA thermoregulation is not driven by the nhba promoter

    To investigate the molecular mechanisms involved in NHBA thermoregulation a nhba

    deletion mutant (Δnhba) was generated in the MC58 strain background by replacement

    with an erythromycin antibiotic resistance cassette and different isogenic

    complementation mutants were generated. To test if the genomic context played a role

    in nhba regulation, the complete sequence, comprising the entire gene and the

    intergenic regulatory region, was inserted into the NMB1428-NMB1429 genomic locus,

    generating the Δnhba-C_nhba strain (Figure 3.5 A). As shown by Western blot analysis

    and qRT-PCR (Figure 3.6 A), the wild type and Δnhba-C_nhba strains showed the same

    expression and thermoregulation of nhba as the wild-type strain, indicating that

    placing the wild type sequence of nhba in another genomic locus does not affect nhba

    regulation in response to temperature changes at both RNA and protein level.

    In order to determine whether the nhba promoter was required for thermoregulation,

    we replaced the MC58 wild-type sequence with an IPTG-inducible Ptac promoter,

    immediately upstream of the CREN sequence (Figure 3.5 B), generating the Δnhba-

    Ptac_nhba strain. We observed an IPTG dose-dependent increase of nhba expression at

    both RNA and protein level. However, using the same amount of IPTG for induction,

    we no longer observed any differences between the transcript levels at the two

    temperatures (Figure 3.6 B). Interestingly, we still observed clear thermoregulation of

    NHBA at protein level, albeit less pronounced than in the wild type strain.

    To determine whether regulatory elements in the nhba upstream intergenic region

    contributed to thermoregulation, we fused the full intergenic region comprising the

    promoter, the CREN sequence and the initial part of the coding sequence

    corresponding to the first 14 amino acids to a mCherry reporter gene (Figure 3.5 C),

    generating the Δnhba-Pwt_mCherry strain. As independent control, we also generated

    Δnhba-Ptac_mCherry strain, a reporter fusion under the control of the IPTG-inducible

    promoter (Figure 3.5 D). We then analyzed the expression of the reporter gene product

    by qRT-PCR and Western blotting of samples collected at either 30°C or 37°C (Figure

    3.6 C). The IPTG inducible promoter drove higher transcription of the downstream

    gene with respect to Pnhba promoter. However, no significant differences were

    observed at protein level. We observed no thermoregulation in either reporter fusion

    construct, both at RNA and at protein level, suggesting that the promoter on its own

    cannot account for the observed differences in expression at the two temperatures. All

  • 39

    together these results highlight that NHBA thermoregulation is at the post-

    transcriptional level.

    Figure 3.5 Schematic representation of nhba mutants generated by ex-locus complementation.

    (A-D) In the MC58Δnhba strain background different mutants were generated by

    complementation in the NMB1428-1429 locus: KanR-kanamycin resistance cassette; CmR-

    chloramphenicol resistance cassette; LacI – LacI repressor gene; Ptac – IPTG inducible promoter;

    mCherry – mCherry reporter gene.

  • 40

    Figure 3.6 NHBA

    thermoregulation is at

    post-transcriptional

    level. (A-C) Wild type

    and recombinant strains

    were grown in GC liquid

    medium at 30°C or 37°C,

    with the indicated

    concentration of IPTG,

    where needed. NHBA

    and mCherry protein

    expression were assessed

    by western blotting by

    using polyclonal mouse

    antisera and monoclonal

    mouse antibody

    (ab167453, abcam),

    respectively (upper

    panel). nhba and mCherry

    RNA steady state levels

    were quantified by qRT-

    PCR and relative

    expression levels were

    determined normalizing

    to 16S-rRNA (lower

    panel).

  • 41

    3.5. Temperature affects nhba RNA half-life

    Transcription produces single-stranded RNA molecules that easily form intra- or

    intermolecular partially double-stranded RNAs, or associate with proteins, which may

    be used to regulate gene expression. To prevent or resolve kinetically trapped

    structures, cells use RNA chaperones that in most cases bind RNA non-specifically to

    help refold RNA or RNA–protein (RNP) complexes in ATP-independent or ATP-

    dependent reactions (Herschlag, D. 1995); (Mohr, S. et al. 2002); (Rajkowitsch, L. et al.

    2007). The kinetic problem of RNA folding is dramatically aggravated at low

    temperatures. Few specific molecular mechanisms have been described to be involved

    in the cold shock response. CspA is the major regulator of the cold-shock response in

    E.coli, where the entire process of cold response is well documented. It is a RNA

    chaperone, able to bind both DNA and RNA, and its own regulation in response to

    temperature downshift is driven by a post-transcriptional mechanism which involves

    the 5’UTR region, resulting in higher transcript stability at low temperatures. The N.

    meningitidis homologue for cspA is NMB0838 and it resulted to be upregulated at 32°C

    in microarray analysis (data not shown). Another class of RNA binding proteins that

    accelerate structural rearrangements of RNA, particularly in the cold, are the DEAD-

    box RNA helicases. By using ATP as substrate, they mediate RNA conformational

    changes, otherwise kinetically unstable (Iost, I. et al. 2013). NMB1368 is a RNA helicase

    H present in N. meningitidis. Therefore, we decided to generate deletion mutants of

    NMB1368 and NMB0838 in MC58 strain background to investigate if this could have

    an effect on NHBA thermoregulation. Mutant strains did not show alterations in

    thermoregulation, both by qRT-PCR and Western blotting (Figure 3.7), even if a

    slightly decrease in expression was observed in ΔNMB0838 mutant grown at 30°C.

  • 42

    Figure 3.7 Deletion of NMB1368 or NMB0838 genes did not affect NHBA thermoregulation.

    NMB1368 or NMB0838 genes were deleted in the MC58 wt strain background. Mutant strains

    were grown in GC broth at 30°C or 37°C until OD600 0.5. Whole cell lysates and samples for

    RNA isolation were collected. (A) Whole cell lysates were prepared and separated by SDS-

    PAGE prior to Western Blotting. The *a symbol indicates a non-specific band used as loading

    control and for relative protein quantification. (B) nhba RNA steady state levels were quantified

    by qRT-PCR and relative expression levels were determined normalizing to 16S-rRNA.

  • 43

    We evaluated the nhba RNA decay after stopping active transcription by addition of

    rifampicin. The relative RNA amount was quantified by qRT-PCR (Figure 3.8). To

    obtain higher accuracy and reliability we used NGH38 as wild-type strain as overall

    nhba expression levels were higher compared to MC58. The transcript of nhba at 37°C

    showed a very short half-life and was rapidly degraded below the limit of detection

    (Figure 3.8). However, nhba transcript decay was found to be directly influenced by

    temperature, showing a shorter half-life at 37°C relative to 30°C

    Figure 3.8 The nhba transcript has a shortened half-life at elevated temperatures. Strain

    NGH38 was grown in GC broth until OD600 0.5 at the defined temperatures. RNA extracts were

    prepared at different time points after active transcription was stopped by adding rifampicin.

    nhba and 16s RNA abundance were measured by qRT-PCR and quantified relatively to the

    levels observed at the start of the experiment. Relative RNA quantity was calculated as 2-(Ct1-Ct0)

    and transformed as Y= log(Y). A linear regression was performed for each dataset and plotted

    as continuous lines. Data represent the mean from three independent biological replicates ± SD.

  • 44

    3.6. NHBA protein shows higher stability at 30°C respect to 37°C

    Having determined that NHBA protein levels are strongly affected post-

    transcriptionally, we wanted to investigate whether altered protein stability at different

    temperatures could account for the observed differences. We therefore grew the MC58

    wild type strain in GC broth at 30°C and 37°C to mid-exponential phase and stopped

    protein translation by adding spectinomycin. Each culture was then split and

    incubated at both 30°C and 37°C and samples for whole cell protein extraction were

    withdrawn at different time-points of treatment. NHBA accumulation upon protein

    translation arrest was analyzed by Western blots with representative results shown in

    Figure 3.9. At 30°C, the amount of NHBA full length protein appears the same for 45-

    60 min treatment (Panel A, upper part), whereas treatment at 37 °C show no change for

    about 10 min treatment (Panel A, lower part).

    Cells grown at 37 °C and treated with spectinomycin at 30 °C show a similar amount

    of NHBA for about 20 min treatment (Panel B, upper part), while treatment at 37 °C

    shows no changes for 10 min (Panel B, lower part). Overall, these data suggest that

    NHBA turnover is higher at 37°C and, accordingly, it appears stable at 30 °C. Thus,

    NHBA thermoregulation is additionally exerted by post-translational mechanisms.

    Figure 3.9 NHBA protein turnover is directly affected by temperature changes. (A-B) MC58

    wild type strain was grown in GC broth until OD600 0.5 at 30°C and 37°C. Whole cell extracts

    were prepared at different timepoints after the active translation was stopped by adding

    spectinomycin. Protein samples were separated by SDS-PAGE prior to Western Blotting using a

    anti-NHBA mouse polyclonal serum. The full length protein is shown. The *a symbol indicates

    a non-specific band used as loading control.

  • 45

    3.7. NHBA expression levels correlate with susceptibility to complement-

    mediated killing by anti-NHBA antibodies

    NHBA is one of the three major components of the 4CMen vaccine against serogroup B

    meningococcus and is a protective antigen that is able to elicit a robust immune

    response. Although NHBA is present in all neisserial species, we have shown that its

    expression is variable among strains and moreover is affected by several factors such

    as like growth phase and temperature changes. It is therefore paramount to

    understand how different NHBA expression levels, either through strain variation or

    triggered by different growth conditions affect the bacterium’s susceptibility to killing

    mediated by anti-NHBA antibodies.

    We therefore investigated whether the different NHBA expression levels at different

    temperatures could affect bacterial susceptibility to complement-mediated killing. To

    address this hypothesis, we grew strain MC58 at both 30°C and 37°C and confirmed

    increased NHBA and decreased fHbp expression levels at 30°C compared to 37°C

    (Figure 3.10 A). Functional antigen should be exposed on the bacterial cell surface and

    we verified that altered antigen expression levels translated into altered surface

    exposure looking at NHBA, fHbp and cps surface exposure by flow cytometry (Figure

    3.10 B). Given these observations, we assessed the ability of immune serum raised

    against NHBA to kill N. meningitidis grown at the two different temperatures by

    determining their serum bactericidal titer (Figure 3.10 C). This assay is also called

    serum bactericidal assay (SBA) and was performed at both 30°C and 37°C. While clear

    differences in antigen expression were evident by Western blotting and flow cytometry

    analysis and trends of the SBA assays reflected these differences, we were unable to

    gain statistical significance due to the intrinsic variability of the experiment. We

    reasoned that altering the temperature would affect many different processes in the

    bacterial cell and confounding pleotropic effects would make it difficult to determine

    the precise impact of NHBA expression levels on serum bactericidal killing. For

    example, important virulence factors such as fHbp and the neisserial capsule also

    respond to temperature changes, but in the opposite way compared to NHBA (Figure

    3.10 B). These pleiotropic effects could then mask the role of NHBA in this assay

    making it impossible to extract how NHBA expression levels affect bacterial killing in

    this assay.

  • 46

    Figure 3.10 Temperature driven expression, surface exposure and susceptibility to

    complement-mediated killing. The MC58 wild type strain was grown at 30°C or 37°C in MH

    broth +0.25% glucose until OD600 0.25 was reached. Bacteria were collected and expression

    levels of NHBA and fHbp were determined by (A) Western blotting and surface exposure of the

    defined antigens was confirmed by (B) flow cytometry using polyclonal antisera. (C) Serum

    bactericidal titers were determined using baby rabbit complement as source of complement

    factors (rabbit SBA, rSBA). rSBA titers indicate the dilution of the α-NHBA mouse polyclonal

    serum, α-fHbp mouse polyclonal serum or α-cps mouse monoclonal antibody at which 50% of

    killing was reached. All the data are representative of three independent biological replicates.

  • 47

    In order to circumvent pleiotropic effect on bacterial expression of virulence factors

    other than NHBA, we generated a recombinant strain in which NHBA expression was

    under the control of an IPTG-inducible promoter (MC58 Δnhba-Ptac_nhba). This assay

    allowed us to perform the experiment under identical conditions while varying NHBA

    expression through addition of different IPTG concentrations. We confirmed protein

    expression and surface exposure of NHBA in these cultures using Western blotting and

    flow cytometry analysis (Figure 3.11 A and B, respectively).This then enabled us to

    extract the role of NHBA expression levels in the ability of anti-NHBA antiserum from

    mice to mediate complement-dependent killing through the rSBA (Figure 3.11 C). We

    observed that rSBA titers correlated directly with NHBA expression levels, as

    confirmed by statistical analysis.

  • 48

    Figure 3.11 Correlation between NHBA expression, surface exposure and susceptibility to complement-mediated killing by anti-NHBA antibodies. MC58

    Δnhba, wild type and Δnhba-Ptac_nhba strains were grown in MH broth +0.25% glucose until OD600 0.25 at 30°C or 37°C as indicated. IPTG was added, where

    needed, at the indicated final concentrations. Bacteria were collected to perform (A) Western blotting, (B) flow cytometry analysis and (C) serum bactericidal assay.

    The relative quantification obtained by (A) densitometry analysis and (B) FACS geometric mean calculation were estimated. All the data represent the mean +/- SD

    from three independent biological replicates and a linear regression was performed on each dataset (black lines indicate the linear fit, dot lines indicate the 95% CI).

    A Pearson correlation test was used to assess the goodness of correlation between the three different datasets (Densitometry/SBA titers: Pearson r = 0,962; P=0.009.

  • 49

    Densitometry/FACS GeoMean: Pearson r = 0,972; P=0.006; FACS GeoMean/SBA titers: Pearson r = 0,925; P=0.008). Western blotting relative quantification of 0.050

    mM IPTG samples were not taken into account for the linear regression as these were found to be out of the linearity range of the assay

  • 50

    3.8. NHBA regulation during invasion

    It has been previously shown that NHBA can be processed by the meningococcal

    protease NalP and human lactoferrin. Cleavage occurs either upstream and

    downstream of the NHBA Arg-rich region resulting in one of two possible cleavage

    fragments termed C2 and C1, respectively (Serruto, D. et al. 2010). To mimic the

    invasive condition MC58 and NGH38 wild type strains were grown until early

    exponential phase and then incubated at 37°C in presence of 25% of human serum

    during a time course, up to 2 hours (Figure 3.12 A and B). After 15 minutes of serum

    incubation NHBA was induced and strongly processed in both strains. In fact, despite

    the appearance of the lower bands typical of the N terminal portion of NHBA after

    cleavage of the protein, the band corresponding to the full length protein appeared to

    retain the same intensity or even more suggesting induction of the full length NHBA as

    well as concomitant processing. Within the blood there is an abundance of components

    with proteolytic activity, so it is not surprising to see an increase in cleavage. The